Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
NPJ Biofilms Microbiomes ; 10(1): 31, 2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38553449

RESUMO

Crosstalk of microbes with human gut epithelia and immune cells is crucial for gut health. However, there is no existing system for a long-term co-culture of human innate immune cells with epithelium and oxygen-intolerant commensal microbes, hindering the understanding of microbe-immune interactions in a controlled manner. Here, we established a gut epithelium-microbe-immune (GuMI) microphysiological system to maintain the long-term continuous co-culture of Faecalibacterium prausnitzii/Faecalibacterium duncaniae with colonic epithelium, antigen-presenting cells (APCs, herein dendritic cells and macrophages), and CD4+ naive T cells circulating underneath the colonic epithelium. In GuMI-APC condition, multiplex cytokine assays suggested that APCs contribute to the elevated level of cytokines and chemokines secreted into both apical and basolateral compartments compared to GuMI condition that lacks APC. In GuMI-APC with F. prausnitzii (GuMI-APC-FP), F. prausnitzii increased the transcription of pro-inflammatory genes such as toll-like receptor 1 (TLR1) and interferon alpha 1 (IFNA1) in the colonic epithelium, without a significant effect on cytokine secretion, compared to the GuMI-APC without bacteria (GuMI-APC-NB). In contrast, in the presence of CD4+ naive T cells (GuMI-APCT-FP), TLR1, IFNA1, and IDO1 transcription levels decreased with a simultaneous increase in F. prausnitzii-induced secretion of pro-inflammatory cytokines (e.g., IL8) compared to GuMI-APC-FP that lacks T cells. These results highlight the contribution of individual innate immune cells in regulating the immune response triggered by the gut commensal F. prausnitzii. The integration of defined populations of immune cells in the gut microphysiological system demonstrated the usefulness of GuMI physiomimetic platform to study microbe-epithelial-immune interactions in healthy and disease conditions.


Assuntos
Faecalibacterium prausnitzii , Sistemas Microfisiológicos , Humanos , Faecalibacterium prausnitzii/fisiologia , Receptor 1 Toll-Like , Citocinas , Inflamação
2.
Res Sq ; 2023 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-37886530

RESUMO

Crosstalk of microbes with human gut epithelia and immune cells is crucial for gut health. However, there is no existing system for a long-term co-culture of human innate immune cells with epithelium and oxygen-intolerant commensal microbes, hindering the understanding of microbe-immune interactions in a controlled manner. Here, we establish a gut epithelium-microbe-immune microphysiological system to maintain the long-term continuous co-culture of Faecalibacterium prausnitzii/Faecalibacterium duncaniae with colonic epithelium, antigen-presenting cells (APCs, herein dendritic cells and macrophages), with CD4+ naïve T cells circulating underneath the colonic epithelium. Multiplex cytokine assays suggested that APCs contribute to the elevated level of cytokines and chemokines being secreted into both apical and basolateral compartments. In contrast, the absence of APCs does not allow reliable detection of these cytokines. In the presence of APCs, F. prausnitzii increased the transcription of pro-inflammatory genes such as toll-like receptor 1 (TLR1) and interferon alpha 1 (IFNA1) in the colonic epithelium, but no significant change on the secreted cytokines. In contrast, integration of CD4+ naïve T cells reverses this effect by decreasing the transcription of TLR1, IFNA1, and indoleamine 2,3-dioxygenase, and increasing the F. prausnitzii-induced secretion of pro-inflammatory cytokines such as IL-8, MCP-1/CCL2, and IL1A. These results highlight the contribution of individual innate immune cells in the regulation of the immune response triggered by the gut commensal F. prausnitzii. The successful integration of defined populations of immune cells in this gut microphysiological system demonstrated the usefulness of the GuMI physiomimetic platform to study microbe-epithelial-immune interactions in health and disease.

3.
FEBS Lett ; 596(5): 681-695, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34923635

RESUMO

Metabolic and inflammatory disorders such as autoimmune and neurodegenerative diseases are increasing at alarming rates. Many of these are not tissue-specific occurrences but complex, systemic pathologies of unknown origin for which no cure exists. Such complexity obscures causal relationships among factors regulating disease progression. Emerging technologies mimicking human physiology, such as microphysiological systems (MPSs), offer new possibilities to provide clarity in systemic metabolic and inflammatory diseases. Controlled interaction of multiple MPSs and the scalability of biological complexity in MPSs, supported by continuous multiomic monitoring, might hold the key to identifying novel relationships between interorgan crosstalk, metabolism, and immunity. In this perspective, I aim to discuss the current state of modeling multiorgan physiology and evaluate current opportunities and challenges.


Assuntos
Fenômenos Fisiológicos Celulares , Dispositivos Lab-On-A-Chip , Humanos
4.
Nat Protoc ; 16(8): 3874-3900, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34183870

RESUMO

The presence of microbes in the colon impacts host physiology. Therefore, microbes are being evaluated as potential treatments for colorectal diseases. Humanized model systems that enable robust culture of primary human intestinal cells with bacteria facilitate evaluation of potential treatments. Here, we describe a protocol that can be used to coculture a primary human colon monolayer with aerotolerant bacteria. Primary human colon cells maintained as organoids are dispersed into single-cell suspensions and then seeded on collagen-coated Transwell inserts, where they attach and proliferate to form confluent monolayers within days of seeding. The confluent monolayers are differentiated for an additional 4 d and then cocultured with bacteria. As an example application, we describe how to coculture differentiated colon cells for 8 h with four strains of Bacteroides thetaiotaomicron, each engineered to detect different colonic microenvironments via genetically embedded logic circuits incorporating deoxycholic acid and anhydrotetracycline sensors. Characterization of this coculture system reveals that barrier function remains intact in the presence of engineered B. thetaiotaomicron. The bacteria stay close to the mucus layer and respond in a microenvironment-specific manner to the inducers (deoxycholic acid and anhydrotetracycline) of the genetic circuits. This protocol thus provides a useful mucosal barrier system to assess the effects of bacterial cells that respond to the colonic microenvironment, and may also be useful in other contexts to model human intestinal barrier properties and microbiota-host interactions.


Assuntos
Bacteroides thetaiotaomicron/fisiologia , Colo/citologia , Células Epiteliais/fisiologia , Mucosa Intestinal/citologia , Técnicas de Cocultura/métodos , Humanos , Organoides
5.
Sci Adv ; 7(5)2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33514545

RESUMO

Slow progress in the fight against neurodegenerative diseases (NDs) motivates an urgent need for highly controlled in vitro systems to investigate organ-organ- and organ-immune-specific interactions relevant for disease pathophysiology. Of particular interest is the gut/microbiome-liver-brain axis for parsing out how genetic and environmental factors contribute to NDs. We have developed a mesofluidic platform technology to study gut-liver-cerebral interactions in the context of Parkinson's disease (PD). It connects microphysiological systems (MPSs) of the primary human gut and liver with a human induced pluripotent stem cell-derived cerebral MPS in a systemically circulated common culture medium containing CD4+ regulatory T and T helper 17 cells. We demonstrate this approach using a patient-derived cerebral MPS carrying the PD-causing A53T mutation, gaining two important findings: (i) that systemic interaction enhances features of in vivo-like behavior of cerebral MPSs, and (ii) that microbiome-associated short-chain fatty acids increase expression of pathology-associated pathways in PD.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doenças Neurodegenerativas , Doença de Parkinson , Encéfalo/metabolismo , Humanos , Fígado/metabolismo , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/metabolismo , Doença de Parkinson/genética , Doença de Parkinson/metabolismo
6.
J Virol ; 95(2)2020 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-33115867

RESUMO

Latent HIV infection is the main barrier to cure, and most HIV-infected cells reside in the gut, where distinct but unknown mechanisms may promote viral latency. Transforming growth factor ß (TGF-ß), which induces the expression of CD103 on tissue-resident memory T cells, has been implicated in HIV latency. Using CD103 as a surrogate marker to identify cells that have undergone TGF-ß signaling, we compared the HIV RNA/DNA contents and cellular transcriptomes of CD103+ and CD103- CD4 T cells from the blood and rectum of HIV-negative (HIV-) and antiretroviral therapy (ART)-suppressed HIV-positive (HIV+) individuals. Like gut CD4+ T cells, circulating CD103+ cells harbored more HIV DNA than did CD103- cells but transcribed less HIV RNA per provirus. Circulating CD103+ cells also shared a gene expression profile that is closer to that of gut CD4 T cells than to that of circulating CD103- cells, with significantly lower expression levels of ribosomal proteins and transcriptional and translational pathways associated with HIV expression but higher expression levels of a subset of genes implicated in suppressing HIV transcription. These findings suggest that blood CD103+ CD4 T cells can serve as a model to study the molecular mechanisms of HIV latency in the gut and reveal new cellular factors that may contribute to HIV latency.IMPORTANCE The ability of HIV to establish a reversibly silent, "latent" infection is widely regarded as the main barrier to curing HIV. Most HIV-infected cells reside in tissues such as the gut, but it is unclear what mechanisms maintain HIV latency in the blood or gut. We found that circulating CD103+ CD4+ T cells are enriched for HIV-infected cells in a latent-like state. Using RNA sequencing (RNA-seq), we found that CD103+ T cells share a cellular transcriptome that more closely resembles that of CD4+ T cells from the gut, suggesting that they are homing to or from the gut. We also identified the cellular genes whose expression distinguishes gut CD4+ or circulating CD103+ T cells from circulating CD103- T cells, including some genes that have been implicated in HIV expression. These genes may contribute to latent HIV infection in the gut and may serve as new targets for therapies aimed at curing HIV.


Assuntos
Antígenos CD/metabolismo , Linfócitos T CD4-Positivos/virologia , Trato Gastrointestinal/virologia , Infecções por HIV/virologia , HIV-1/fisiologia , Cadeias alfa de Integrinas/metabolismo , Transcrição Gênica/genética , Antivirais/uso terapêutico , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , DNA Viral/metabolismo , Trato Gastrointestinal/imunologia , Regulação da Expressão Gênica , Infecções por HIV/tratamento farmacológico , Humanos , Linfócitos Intraepiteliais/metabolismo , Linfócitos Intraepiteliais/virologia , Provírus/fisiologia , RNA Viral/metabolismo , Proteínas Ribossômicas/genética , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/virologia , Latência Viral
7.
Cell Syst ; 10(3): 223-239.e9, 2020 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-32191873

RESUMO

Although the association between the microbiome and IBD and liver diseases is known, the cause and effect remain elusive. By connecting human microphysiological systems of the gut, liver, and circulating Treg and Th17 cells, we created a multi-organ model of ulcerative colitis (UC) ex vivo. The approach shows microbiome-derived short-chain fatty acids (SCFAs) to either improve or worsen UC severity, depending on the involvement of effector CD4 T cells. Using multiomics, we found SCFAs increased production of ketone bodies, glycolysis, and lipogenesis, while markedly reducing innate immune activation of the UC gut. However, during acute T cell-mediated inflammation, SCFAs exacerbated CD4+ T cell-effector function, partially through metabolic reprograming, leading to gut barrier disruption and hepatic injury. These paradoxical findings underscore the emerging utility of human physiomimetic technology in combination with systems immunology to study causality and the fundamental entanglement of immunity, metabolism, and tissue homeostasis.


Assuntos
Ácidos Graxos Voláteis/metabolismo , Trato Gastrointestinal/metabolismo , Fígado/metabolismo , Biomimética/métodos , Microbioma Gastrointestinal/fisiologia , Homeostase , Humanos , Inflamação/metabolismo , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/fisiopatologia , Mucosa Intestinal/metabolismo , Modelos Biológicos , Linfócitos T Reguladores/imunologia , Células Th17/imunologia
8.
ALTEX ; 37(3): 365-394, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32113184

RESUMO

The first microfluidic microphysiological systems (MPS) entered the academic scene more than 15 years ago and were considered an enabling technology to human (patho)biology in vitro and, therefore, provide alternative approaches to laboratory animals in pharmaceutical drug development and academic research. Nowadays, the field generates more than a thousand scientific publications per year. Despite the MPS hype in academia and by platform providers, which says this technology is about to reshape the entire in vitro culture landscape in basic and applied research, MPS approaches have neither been widely adopted by the pharmaceutical industry yet nor reached regulated drug authorization processes at all. Here, 46 leading experts from all stakeholders - academia, MPS supplier industry, pharmaceutical and consumer products industries, and leading regulatory agencies - worldwide have analyzed existing challenges and hurdles along the MPS-based assay life cycle in a second workshop of this kind in June 2019. They identified that the level of qualification of MPS-based assays for a given context of use and a communication gap between stakeholders are the major challenges for industrial adoption by end-users. Finally, a regulatory acceptance dilemma exists against that background. This t4 report elaborates on these findings in detail and summarizes solutions how to overcome the roadblocks. It provides recommendations and a roadmap towards regulatory accepted MPS-based models and assays for patients' benefit and further laboratory animal reduction in drug development. Finally, experts highlighted the potential of MPS-based human disease models to feedback into laboratory animal replacement in basic life science research.


Assuntos
Alternativas aos Testes com Animais , Bem-Estar do Animal , Desenvolvimento de Medicamentos , Avaliação Pré-Clínica de Medicamentos/métodos , Dispositivos Lab-On-A-Chip , Animais , Indústria Farmacêutica , Humanos , Modelos Biológicos
9.
Nat Commun ; 10(1): 4344, 2019 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-31554802

RESUMO

Innate immune responses to Zika virus (ZIKV) are dampened in the lower female reproductive tract (LFRT) compared to other tissues, but the mechanism that underlies this vulnerability is poorly understood. Using tissues from uninfected and vaginally ZIKV-infected macaques and mice, we show that low basal expression of RNA-sensing pattern recognition receptors (PRRs), or their co-receptors, in the LFRT contributes to high viral replication in this tissue. In the LFRT, ZIKV sensing provides limited protection against viral replication, and the sensors are also minimally induced after vaginal infection. While IFNα/ß receptor signaling offers minimal protection in the LFRT, it is required to prevent dissemination of ZIKV to other tissues, including the upper FRT. Our findings support a role for RNA-sensing PRRs in the dampened innate immunity against ZIKV in the LFRT compared to other tissues and underlie potential implications for systemic dissemination upon heterosexual transmission of ZIKV in women.


Assuntos
Genitália Feminina/imunologia , Imunidade Inata/imunologia , RNA Viral/imunologia , Infecção por Zika virus/imunologia , Zika virus/imunologia , Animais , Feminino , Regulação Viral da Expressão Gênica , Genitália Feminina/metabolismo , Genitália Feminina/virologia , Humanos , Imunidade Inata/genética , Macaca mulatta , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Viral/genética , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/imunologia , Receptor de Interferon alfa e beta/metabolismo , Receptores de Reconhecimento de Padrão/genética , Receptores de Reconhecimento de Padrão/imunologia , Receptores de Reconhecimento de Padrão/metabolismo , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/imunologia , Receptor 3 Toll-Like/metabolismo , Vagina/imunologia , Vagina/metabolismo , Vagina/virologia , Replicação Viral/genética , Replicação Viral/imunologia , Zika virus/genética , Zika virus/fisiologia , Infecção por Zika virus/genética , Infecção por Zika virus/virologia
10.
PLoS Pathog ; 14(2): e1006856, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29470552

RESUMO

HIV-1-infected cells persist indefinitely despite the use of combination antiretroviral therapy (ART), and novel therapeutic strategies to target and purge residual infected cells in individuals on ART are urgently needed. Here, we demonstrate that CD4+ T cell-associated HIV-1 RNA is often highly enriched in cells expressing CD30, and that cells expressing this marker considerably contribute to the total pool of transcriptionally active CD4+ lymphocytes in individuals on suppressive ART. Using in situ RNA hybridization studies, we show co-localization of CD30 with HIV-1 transcriptional activity in gut-associated lymphoid tissues. We also demonstrate that ex vivo treatment with brentuximab vedotin, an antibody-drug conjugate (ADC) that targets CD30, significantly reduces the total amount of HIV-1 DNA in peripheral blood mononuclear cells obtained from infected, ART-suppressed individuals. Finally, we observed that an HIV-1-infected individual, who received repeated brentuximab vedotin infusions for lymphoma, had no detectable virus in peripheral blood mononuclear cells. Overall, CD30 may be a marker of residual, transcriptionally active HIV-1 infected cells in the setting of suppressive ART. Given that CD30 is only expressed on a small number of total mononuclear cells, it is a potential therapeutic target of persistent HIV-1 infection.


Assuntos
Linfócitos T CD4-Positivos/virologia , Infecções por HIV/virologia , HIV-1/fisiologia , Antígeno Ki-1/metabolismo , Tecido Linfoide/virologia , Reto/virologia , Ativação Transcricional , Fármacos Anti-HIV/farmacologia , Terapia Antirretroviral de Alta Atividade , Biomarcadores/sangue , Biomarcadores/metabolismo , Brentuximab Vedotin , Contagem de Linfócito CD4 , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Células Cultivadas , Estudos de Coortes , DNA Viral/sangue , DNA Viral/metabolismo , Infecções por HIV/tratamento farmacológico , Infecções por HIV/metabolismo , Infecções por HIV/patologia , HIV-1/efeitos dos fármacos , HIV-1/isolamento & purificação , HIV-1/patogenicidade , Humanos , Imunoconjugados/farmacologia , Hibridização In Situ , Antígeno Ki-1/antagonistas & inibidores , Antígeno Ki-1/sangue , Antígeno Ki-1/química , Tecido Linfoide/efeitos dos fármacos , Tecido Linfoide/metabolismo , Tecido Linfoide/patologia , RNA Viral/sangue , RNA Viral/metabolismo , Reto/efeitos dos fármacos , Reto/metabolismo , Reto/patologia , Solubilidade , Ativação Transcricional/efeitos dos fármacos , Carga Viral/efeitos dos fármacos
11.
Mucosal Immunol ; 11(4): 1158-1167, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29456247

RESUMO

Determining the magnitude of local immune response during mucosal exposure to viral pathogens is critical to understanding the mechanism of viral pathogenesis. We previously showed that vaginal inoculation of lymphocytic choriomeningitis virus (LCMV) fails to induce a robust innate immune response in the lower female reproductive tract (FRT), allowing high titer viral replication and a delay in T-cell-mediated viral control. Despite this immunological delay, LCMV replication remained confined mainly to the FRT and the draining iliac lymph node. Here, we show that rectal infection with LCMV triggers type I/III interferon responses, followed by innate immune activation and lymphocyte recruitment to the colon. In contrast to vaginal exposure, innate immunity controls LCMV replication in the colon, but virus rapidly disseminates systemically. Virus-induced inflammation promotes the recruitment of LCMV target cells to the colon followed by splenic viral dissemination by infected B cells, and to a lesser extent by CD8 T cells. These findings demonstrate major immunological differences between vaginal and rectal exposure to the same viral pathogen, highlighting unique risks associated with each of these common routes of sexual viral transmission.


Assuntos
Infecções por Arenaviridae/imunologia , Linfócitos B/imunologia , Colo/imunologia , Linfócitos/imunologia , Vírus da Coriomeningite Linfocítica/fisiologia , Vagina/imunologia , Animais , Linfócitos B/virologia , Movimento Celular , Colo/virologia , Feminino , Imunidade Inata , Ativação Linfocitária , Vírus da Coriomeningite Linfocítica/patogenicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reto/metabolismo , Vagina/virologia
12.
AIDS Res Hum Retroviruses ; 33(S1): S31-S39, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28882052

RESUMO

The gastrointestinal (GI) tract harbors most of the body's immune cells and is also a major HIV reservoir in ART-treated patients. To achieve a cure, most HIV-infected cells must be identified and eliminated. While obtaining gut biopsies is a relatively noninvasive method of sampling relevant tissue for monitoring HIV activity, immune cell isolation from these limited tissue samples has proven to be challenging. Enzymatic tissue digestion is required for maximal immune cell isolation from gut biopsies. However, these enzymatic digestions can also be detrimental for preservation of cellular surface markers that are required for accurate identification of various subsets of leukocytes. In this study, we describe an optimized protocol for isolation of lymphocytes from human gut biopsies. We also discuss our validation results, which show that compared with several other collagenase preparations, the use of CSLPA maintains high lymphocyte recovery while preserving the integrity of most cellular surface antigens that we tested. Importantly, chemokine receptors that are used to characterize various subsets of T cells, which are notorious for being digested during a typical enzymatic tissue digestion, are highly preserved using this protocol.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Separação Celular/métodos , Trato Gastrointestinal/imunologia , Infecções por HIV/imunologia , Biópsia/métodos , Contagem de Linfócito CD4 , Quimiocinas/análise , Trato Gastrointestinal/virologia , HIV-1/imunologia , Humanos
13.
AIDS Res Hum Retroviruses ; 33(S1): S40-S58, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28882067

RESUMO

The single greatest challenge to an HIV cure is the persistence of latently infected cells containing inducible, replication-competent proviral genomes, which constitute only a small fraction of total or infected cells in the body. Although resting CD4+ T cells in the blood are a well-known source of viral rebound, more than 90% of the body's lymphocytes reside elsewhere. Many are in gut tissue, where HIV DNA levels per million CD4+ T cells are considerably higher than in the blood. Despite the significant contribution of gut tissue to viral replication and persistence, little is known about the cell types that support persistence of HIV in the gut; importantly, T cells in the gut have phenotypic, functional, and survival properties that are distinct from T cells in other tissues. The mechanisms by which latency is established and maintained will likely depend on the location and cytokine milieu surrounding the latently infected cells in each compartment. Therefore, successful HIV cure strategies require identification and characterization of the exact cell types that support viral persistence, particularly in the gut. In this review, we describe the seeding of the latent HIV reservoir in the gut mucosa; highlight the evidence for compartmentalization and depletion of T cells; summarize the immunologic consequences of HIV infection within the gut milieu; propose how the damaged gut environment may promote the latent HIV reservoir; and explore several immune cell targets in the gut and their place on the path toward HIV cure.


Assuntos
Linfócitos T CD4-Positivos/virologia , Infecções por HIV/imunologia , Tecido Linfoide/imunologia , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Células Th17/imunologia , Latência Viral/imunologia , Fármacos Anti-HIV/uso terapêutico , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , HIV-1/imunologia , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/virologia , Tecido Linfoide/citologia , Replicação Viral/imunologia
14.
J Exp Med ; 213(13): 2913-2929, 2016 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-27852793

RESUMO

Understanding the host immune response to vaginal exposure to RNA viruses is required to combat sexual transmission of this class of pathogens. In this study, using lymphocytic choriomeningitis virus (LCMV) and Zika virus (ZIKV) in wild-type mice, we show that these viruses replicate in the vaginal mucosa with minimal induction of antiviral interferon and inflammatory response, causing dampened innate-mediated control of viral replication and a failure to mature local antigen-presenting cells (APCs). Enhancement of innate-mediated inflammation in the vaginal mucosa rescues this phenotype and completely inhibits ZIKV replication. To gain a better understanding of how this dampened innate immune activation in the lower female reproductive tract may also affect adaptive immunity, we modeled CD8 T cell responses using vaginal LCMV infection. We show that the lack of APC maturation in the vaginal mucosa leads to a delay in CD8 T cell activation in the draining lymph node and hinders the timely appearance of effector CD8 T cells in vaginal mucosa, thus further delaying viral control in this tissue. Our study demonstrates that vaginal tissue is exceptionally vulnerable to infection by RNA viruses and provides a conceptual framework for the male to female sexual transmission observed during ZIKV infection.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/fisiologia , Vagina/imunologia , Replicação Viral/imunologia , Infecção por Zika virus/imunologia , Zika virus/fisiologia , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/patologia , Linfócitos T CD8-Positivos/patologia , Feminino , Imunidade Inata , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/patologia , Camundongos , Camundongos Knockout , Vagina/patologia , Vagina/virologia , Replicação Viral/genética , Infecção por Zika virus/genética , Infecção por Zika virus/patologia
15.
Sci Rep ; 6: 34373, 2016 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-27687492

RESUMO

Corneal wound healing is often affected by TGF-ß-mediated fibrosis and scar formation. Guided fibrosis with IGF-1 and antifibrotic substances might maintain corneal transparency. Primary human corneal keratocytes under serum-free conditions were used as a model of corneal stromal wounding, with markers of corneal fibrosis and opacity studied under TGF-ß2 stimulation. Single-cell imaging flow cytometry was used to determine nuclearization of Smad3, and intracellular fluorescence intensity of Smad7 and the corneal crystallin aldehyde dehydrogenase 3A1. Extracellular matrix proteoglycans keratocan and biglycan were quantified using ELISAs. On the TGF-ß2 background, the keratocytes were treated with IGF-1, and suberoylanilidehydroxamic acid (SAHA) or halofuginone ± IGF-1. IGF-1 alone decreased Smad3 nuclearization and increased aldehyde dehydrogenase 3A1 expression, with favorable extracellular matrix proteoglycan composition. SAHA induced higher Smad7 levels and inhibited translocation of Smad3 to the nucleus, also when combined with IGF-1. Immunofluorescence showed that myofibroblast transdifferentiation is attenuated and appearance of fibroblasts is favored by IGF-1 alone and in combination with the antifibrotic substances. The TGF-ß/Smad pathway of fibrosis and opacity was inhibited by IGF-1, and further with SAHA in particular, and with halofuginone. IGF-1 is thus a valid aid to antifibrotic treatment, with potential for effective and transparent corneal wound healing.

16.
Wien Klin Wochenschr ; 127 Suppl 5: S204-9, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25821058

RESUMO

The intestinal epithelium is composed of diverse cell types, most abundant being the enterocytes. Among other functions, they maintain the intestinal barrier and play a critical role in the absorption of nutrients, drugs and toxins. This study describes the development and characterization of human intestinal epithelial cells (HUIEC), a spontaneously arising cell line established by selective trypsinization and cloning of the intestinal epithelium, resulting in a uniform population of highly epithelial cells with a strong growth potential.


Assuntos
Técnicas de Cultura Celular por Lotes/métodos , Separação Celular/métodos , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Mucosa Intestinal/citologia , Diferenciação Celular/fisiologia , Linhagem Celular , Células Epiteliais/classificação , Humanos
17.
PLoS One ; 10(2): e0117302, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25679956

RESUMO

Regeneration of skeletal muscle after injury is limited by scar formation, slow healing time and a high recurrence rate. A therapy based on platelet-rich plasma (PRP) has become a promising lead for tendon and ligament injuries in recent years, however concerns have been raised that PRP-derived TGF-ß could contribute to fibrotic remodelling in skeletal muscle after injury. Due to the lack of scientific grounds for a PRP -based muscle regeneration therapy, we have designed a study using human myogenic progenitors and evaluated the potential of PRP alone and in combination with decorin (a TGF-ß inhibitor), to alter myoblast proliferation, metabolic activity, cytokine profile and expression of myogenic regulatory factors (MRFs). Advanced imaging multicolor single-cell analysis enabled us to create a valuable picture on the ratio of quiescent, activated and terminally committed myoblasts in treated versus control cell populations. Finally high-resolution confocal microscopy validated the potential of PRP and decorin to stimulate the formation of polynucleated myotubules. PRP was shown to down-regulate fibrotic cytokines, increase cell viability and proliferation, enhance the expression of MRFs, and contribute to a significant myogenic shift during differentiation. When combined with decorin further synergistc effects were identified. These results suggest that PRP could not only prevent fibrosis but could also stimulate muscle commitment, especially when combined with a TGF-ß inhibitor.


Assuntos
Diferenciação Celular , Desenvolvimento Muscular , Mioblastos/citologia , Mioblastos/metabolismo , Plasma Rico em Plaquetas , Fator de Crescimento Transformador beta/antagonistas & inibidores , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Decorina/metabolismo , Decorina/farmacologia , Desmina/genética , Desmina/metabolismo , Humanos , Desenvolvimento Muscular/efeitos dos fármacos , Desenvolvimento Muscular/genética , Mioblastos/efeitos dos fármacos , Miostatina/genética , Miostatina/metabolismo
18.
J Muscle Res Cell Motil ; 35(5-6): 249-57, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25380573

RESUMO

Research on skeletal muscles suffers from a lack of appropriate human models to study muscle formation and regeneration on the regulatory level of single cells. This hampers both basic understanding and the development of new therapeutic approaches. The use of imaging multicolour flow cytometry and myogenic stem cells can help fill this void by allowing researchers to visualize and quantify the reaction of individual cultured cells to bioactives or other physiological impulses. As proof of concept, we subjected human CD56+ satellite cells to reference bioactives follistatin and Malva sylvestris extracts and then used imaging multicolor flow cytometry to visualize the stepwise activation of myogenic factors MyoD and myogenin in individual cells. This approach enabled us to evaluate the potency of these bioactives to stimulate muscle commitment. To validate this method, we used multi-photon confocal microscopy to confirm the potential of bioactives to stimulate muscle differentiation and expression of desmin. Imaging multicolor flow cytometry revealed statistically significant differences between treated and untreated groups of myogenic progenitors and we propose the utilization of this concept as an integral part of future muscle research strategies.


Assuntos
Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/citologia , Células-Tronco/citologia , Antígeno CD56/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Desmina/biossíntese , Citometria de Fluxo/métodos , Folistatina/farmacologia , Humanos , Imuno-Histoquímica , Malva/química , Microscopia Confocal , Proteínas Musculares/metabolismo , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Proteína MyoD/metabolismo , Miogenina/metabolismo , Extratos Vegetais/farmacologia , Células Satélites de Músculo Esquelético/efeitos dos fármacos , Células Satélites de Músculo Esquelético/metabolismo , Análise de Célula Única/métodos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
19.
New Microbiol ; 37(1): 51-64, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24531171

RESUMO

Plasma lipid levels are important risk factors for the development of atherosclerosis and coronary heart disease. Previous findings have shown that probiotic bacteria exert positive effects on hypercholesterolemia by lowering serum cholesterol and improving lipid profile that, in turn, leads to a reduced risk of coronary heart disease and atherosclerosis. Most of these studies were carried out with tumoral cell lines that have a metabolism quite different from that of normal cells and may thus respond differently to various stimuli. Here, we demonstrate the beneficial effects of some probiotics on cholesterol levels and pathways in normal small intestinal foetal epithelial tissue cells. The results show that Lactobacillus plantarum strain PCS 26 efficiently removes cholesterol from media, exhibits bile salt hydrolase activity, and up-regulates several genes involved in cholesterol metabolism. This study suggests that Lactobacillus plantarum PCS 26 might act as a liver X receptor agonist and help to improve lipid profiles in hypercholesterolemic patients or even dislipidemias in complex diseases such as the metabolic syndrome.


Assuntos
Colesterol/metabolismo , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Lactobacillus plantarum/metabolismo , Probióticos/metabolismo , Expressão Gênica , Humanos , Lactobacillus plantarum/crescimento & desenvolvimento , Modelos Biológicos , Proteínas/genética , Proteínas/metabolismo
20.
PLoS One ; 9(1): e86297, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24454965

RESUMO

The early establishment of a complete microbiome has been shown to play an integral part in the development and maintenance of an intact intestine and its immune system, although much remains unknown about the specific mechanisms of immune modulation in newborns. In our study we show in a co-culture model of the undeveloped small intestine that members of Lactobacillus spp. influence STAT1 and NF-kB p65 nuclear translocation in both intestinal epithelial cells as well as underlying macrophages. Moreover, by using imaging flow cytometry we were able to monitor each individual cell and create a framework of the percentage of cells in which translocation occurred in challenged versus control cell populations. We also observed a significant difference in baseline translocation in intestinal cells when cultured alone versus those in a co-culture model, underpinning the importance of 3D models over monolayer set-ups in epithelial in vitro research. In conclusion, our work offers new insights into the potential routes by which the commensal microbiome primes the early immune system to fight pathogens, and shows how strain-specific these mechanisms really are.


Assuntos
Enterócitos/imunologia , Imunomodulação , Lactobacillus/fisiologia , Macrófagos/imunologia , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição RelA/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Técnicas de Cocultura , Humanos , Tolerância Imunológica , Intestino Delgado/citologia , Intestino Delgado/imunologia , Intestino Delgado/microbiologia , Lactobacillus/imunologia , Microbiota/imunologia , Transporte Proteico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA